Alcohol dependence is a highly prevalent and debilitating disorder that exhibits relapse rates over 80% despite current treatment efforts. Human alcoholics as well as animal models of dependence exhibit chronic alterations in gene expression that underlie neuroplastic alterations within the reward circuitry of the brain. The mechanisms that lead to chronic disruption of gene expression profiles remain unclear. Alterations in epigenetic modifications to DNA and histone tails have been shown to be associated with alcohol dependence. These epigenetic changes could underlie chronic alterations in gene expression, cellular dysfunction, and ultimately contribute to alcohol seeking behaviors. However, the epigenetic regulatory pathways upstream of these changes remain to be explored. Transcriptomic analysis identified two epigenetic enzymes that are dysregulated in key brain reward regions of a well validated post-dependent (PD) rodent model of alcohol dependence, PR domain containing 2 (PRDM2) and lysine-specific demethylase 6B (KDM6B). These enzymes have both been shown to modulate NF-?B-mediated inflammatory responses in cultured macrophages and microglia. Viral-mediated in vivo shRNA knockdown of PRDM2 in the prefrontal cortex increased alcohol seeking behaviors. The known roles of PRDM2 and KDM6B in immune cell responses are consistent with evidence that inflammatory signaling pathways are critical for the development of uncontrolled drinking behaviors. A combination of behavioral, epigenomic, transcriptomic, and histological approaches will be used to test whether PRDM2 and KDM6B mediate epigenetic control of inflammation-associated transcriptional changes that underlie alcohol dependence. The expression of PRDM2 and KDM6B will first be manipulated using viral vectors delivered to brain reward regions in order to characterize their effects on alcohol-seeking behaviors compared to assessments of general locomotion, anxiety, and natural reward. Chromatin immunoprecipitation followed by DNA sequencing (ChIP-seq), will be used to identify the gene loci subject to dynamic regulation by PRDM2 and KDM6B in alcohol dependent rats. [The effects of PRDM2 and KDM6B are on microglial activation, NF-?B activation, and inflammatory cytokine expression will be functionally assessed using histological, pharmacological, and biochemical analysis in PD rats.] These studies have potential to identify novel epigenetic mechanisms that underlie long-term transcriptional dysregulation, inflammatory signaling, and maladaptive drinking behaviors in alcohol dependence. Given the emerging interest in targeting epigenetic enzymes using small molecule therapies for complex disease, the identification of epigenetic signaling networks that are dysregulated in alcohol dependence could lead to novel therapeutic approaches for this intractable disorder.

Public Health Relevance

Alcohol dependence is a debilitating disorder characterized by long-term changes in brain function that underlie pathological drinking behaviors. We demonstrated that epigenetic enzymes, which are known to mediate long-term changes in cellular function, contribute to alcohol dependence when their expression is manipulated in the brain of rodents. The goal of the proposed studies is to determine the mechanisms by which these epigenetic enzymes reprogram the brain for alcohol dependence, knowledge that may ultimately lead to novel therapeutic strategies.

Agency
National Institute of Health (NIH)
Institute
National Institute on Alcohol Abuse and Alcoholism (NIAAA)
Type
Research Project (R01)
Project #
1R01AA023781-01A1
Application #
9030357
Study Section
Health Services Research Review Subcommittee (AA)
Program Officer
Reilly, Matthew
Project Start
2015-09-24
Project End
2020-08-31
Budget Start
2015-09-24
Budget End
2016-08-31
Support Year
1
Fiscal Year
2015
Total Cost
$345,375
Indirect Cost
$120,375
Name
University of Miami School of Medicine
Department
Psychiatry
Type
Schools of Medicine
DUNS #
052780918
City
Coral Gables
State
FL
Country
United States
Zip Code
33146
Swords, Ronan T; Azzam, Diana; Al-Ali, Hassan et al. (2018) Ex-vivo sensitivity profiling to guide clinical decision making in acute myeloid leukemia: A pilot study. Leuk Res 64:34-41
Lohse, Ines; Al-Ali, Hassan; Volmar, Claude-Henry et al. (2018) Ex vivo drug sensitivity testing as a means for drug repurposing in esophageal adenocarcinoma. PLoS One 13:e0203173
Yang, Hui; Kurtenbach, Stefan; Guo, Ying et al. (2018) Gain of function of ASXL1 truncating protein in the pathogenesis of myeloid malignancies. Blood 131:328-341
Mustafi, Sushmita; Camarena, Vladimir; Volmar, Claude-Henry et al. (2018) Vitamin C Sensitizes Melanoma to BET Inhibitors. Cancer Res 78:572-583
Khorkova, Olga; Wahlestedt, Claes (2017) Oligonucleotide therapies for disorders of the nervous system. Nat Biotechnol 35:249-263
Sartor, Gregory C; Powell, Samuel K; Velmeshev, Dmitry et al. (2017) Cocaine alters Homer1 natural antisense transcript in the nucleus accumbens. Mol Cell Neurosci 85:183-189
Volmar, Claude-Henry; Salah-Uddin, Hasib; Janczura, Karolina J et al. (2017) M344 promotes nonamyloidogenic amyloid precursor protein processing while normalizing Alzheimer's disease genes and improving memory. Proc Natl Acad Sci U S A 114:E9135-E9144
Barbier, E; Johnstone, A L; Khomtchouk, B B et al. (2017) Dependence-induced increase of alcohol self-administration and compulsive drinking mediated by the histone methyltransferase PRDM2. Mol Psychiatry 22:1746-1758
Jones, Candace; Barrera, Ingrid; Brothers, Shaun et al. (2017) Oxytocin and social functioning. Dialogues Clin Neurosci 19:193-201
Khomtchouk, Bohdan B; Hennessy, James R; Wahlestedt, Claes (2017) shinyheatmap: Ultra fast low memory heatmap web interface for big data genomics. PLoS One 12:e0176334

Showing the most recent 10 out of 15 publications